Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Neurosci ; 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38951035

RESUMO

Sleep control depends on a delicate interplay among brain regions. This generates a complex temporal architecture with numerous sleep-stage transi-tions and intermittent fluctuations to micro-states and brief arousals within sleep stages. These temporal dynamics exhibit hallmarks of criticality, suggest-ing that tuning to criticality is essential for spontaneous sleep-stage and arousal transitions. However, how the brain maintains criticality remains not under-stood. Here, we investigate dynamics of θ- and δ-bursts during the sleep-wake cycle of rats (Sprague-Dawley, adult male) with lesion in the wake-promoting locus coeruleus (LC). We show that, in control rats, θ- and δ-bursts exhibit duality of power-law (θ-bursts, active phase) and exponential-like (δ-bursts, quiescent phase) duration distributions, as well as power-law long-range tem-poral correlations (LRTC)-typical of non-equilibrium systems self-organizing at criticality. Further, consecutive θ- and δ-bursts durations are characterized by anti-correlated coupling, indicating a new class of self-organized critical- ity that emerges from underlying feedback between neuronal populations and brain areas involved in generating arousals and sleep states. In contrast, we uncover that LC lesion leads to alteration of θ- and δ-burst critical features, with change in duration distributions and correlation properties, and increase in θ-δ coupling. Notably, these LC-lesion effects are opposite to those observed for lesions in the sleep-promoting ventrolateral preoptic nucleus (VLPO). Our findings indicate that critical dynamics of θ- and δ-bursts arise from a bal-anced interplay of LC and VLPO, which maintains brain tuning to criticality across the sleep-wake cycle-a continuous non-equilibrium behavior in sleepSignificance statement Criticality has been associated with healthy brain function in both sleep and wake. However, how the sleep-wake control circuitry maintains criticality remains not un-derstood. Our analyses demonstrate that arousal promoting neurons in the LC play a key role in maintaining brain criticality across the sleep-wake cycle. The results show that lesions of the wake-promoting LC affect the critical dynamics of θ and δ bursts, altering duration distributions, correlation properties, and θ-δ coupling. The reported changes in criticality measures are opposite to those caused by lesions of the sleep-promoting VLPO. This suggests that feed-forward and feedback interactions among neuronal populations in the LC and VLPO are essential to maintain the brain tuned to criticality across the sleep-wake cycle.

2.
J Neurosci ; 39(25): 4986-4998, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31036764

RESUMO

Most brain neurons are active in waking, but hypothalamic neurons that synthesize the neuropeptide melanin-concentrating hormone (MCH) are claimed to be active only during sleep, particularly rapid eye movement (REM) sleep. Here we use deep-brain imaging to identify changes in fluorescence of the genetically encoded calcium (Ca2+) indicator GCaMP6 in individual hypothalamic neurons that contain MCH. An in vitro electrophysiology study determined a strong relationship between depolarization and Ca2+ fluorescence in MCH neurons. In 10 freely behaving MCH-cre mice (male and female), the highest fluorescence occurred in all recorded neurons (n = 106) in REM sleep relative to quiet waking or non-REM sleep. Unexpectedly, 70% of the MCH neurons had strong fluorescence activity when the mice explored novel objects. Spatial and temporal mapping of the change in fluorescence between pairs of MCH neurons revealed dynamic activation of MCH neurons during REM sleep and activation of a subset of the same neurons during exploratory behavior. Functional network activity maps will facilitate comparisons of not only single-neuron activity, but also network responses in different conditions and disease.SIGNIFICANCE STATEMENT Functional activity maps identify brain circuits responding to specific behaviors, including rapid eye movement sleep (REM sleep), a sleep phase when the brain is as active as in waking. To provide the first activity map of individual neurons during REM sleep, we use deep-brain calcium imaging in unrestrained mice to map the activity of hypothalamic melanin-concentrating hormone (MCH) neurons. MCH neurons were found to be synchronously active during REM sleep, and also during the exploration of novel objects. Spatial mapping revealed dynamic network activation during REM sleep and activation of a subset of the neurons during exploratory behavior. Functional activity maps at the cellular level in specific behaviors, including sleep, are needed to establish a brain connectome.


Assuntos
Comportamento Exploratório/fisiologia , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Mapeamento Encefálico , Cálcio/metabolismo , Feminino , Masculino , Camundongos , Imagem Óptica
3.
Eur J Neurosci ; 43(10): 1298-306, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26369866

RESUMO

A distributed network of neurons regulates wake, non-rapid eye movement (NREM) sleep, and REM sleep. However, there are also glia in the brain, and there is growing evidence that neurons and astroglia communicate intimately to regulate behaviour. To identify the effect of optogenetic stimulation of astrocytes on sleep, the promoter for the astrocyte-specific cytoskeletal protein, glial fibrillary acidic protein (GFAP) was used to direct the expression of channelrhodopsin-2 (ChR2) and the linked reporter gene, enhanced yellow fluorescent protein (EYFP), in astrocytes. rAAV-GFAP-ChR2 (H134R)-EYFP or rAAV-GFAP-EYFP was microinjected (750 nL) into the posterior hypothalamus (bilateral) of mice. Three weeks later baseline sleep was recorded (0 Hz) and 24 h later optogenetic stimulation applied during the first 6 h of the lights-off period. Mice with ChR2 were given 5, 10 or 30 Hz stimulation for 6 h (10-ms pulses; 1 mW; 1 min on 4 min off). At least 36 h elapsed between the stimulation periods (5, 10, 30 Hz) and although 0 Hz was always first, the order of the other three stimulation rates was randomised. In mice with ChR2 (n = 7), 10 Hz, but not 5 or 30 Hz stimulation increased both NREM and REM sleep during the 6-h period of stimulation. Delta power did not increase. In control mice (no ChR2; n = 5), 10 Hz stimulation had no effect. This study demonstrates that direct stimulation of astrocytes powerfully induces sleep during the active phase of the sleep-wake cycle and underlines the inclusion of astrocytes in network models of sleep-wake regulation.


Assuntos
Astrócitos/fisiologia , Hipotálamo Posterior/fisiologia , Optogenética , Sono , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sono REM
4.
Eur J Neurosci ; 43(5): 681-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26741960

RESUMO

Narcolepsy is a chronic sleep disorder linked to the loss of orexin-producing neurons in the hypothalamus. Cataplexy, a sudden loss of muscle tone during waking, is an important distinguishing symptom of narcolepsy and it is often triggered by strong emotions. The neural circuit underlying cataplexy attacks is not known, but is likely to involve the amygdala, a region implicated in regulating emotions. In mice models of narcolepsy, transfer of the orexin gene into surrogate neurons has been successful in ameliorating narcoleptic symptoms. However, it is not known whether this method also blocks cataplexy triggered by strong emotions. To examine this possibility, the gene encoding mouse prepro-orexin was transferred into amygdala neurons of orexin-knockout (KO) mice (rAAV-orexin; n = 8). Orexin-KO mice that did not receive gene transfer (no-rAAV; n = 7) or received only the reporter gene (rAAV-GFP; n = 7) served as controls. Three weeks later, the animal's sleep and behaviour were recorded at night (no-odour control night), followed by another recording at night in the presence of predator odour (odour night). Orexin-KO mice given the orexin gene transfer into surrogate amygdala neurons had significantly less spontaneous bouts of cataplexy, and predator odour did not induce cataplexy compared with control mice. Moreover, the mice with orexin gene transfer were awake more during the odour night. These results demonstrate that orexin gene transfer into amygdala neurons can suppress both spontaneous and emotion-induced cataplexy attacks in narcoleptic mice. It suggests that manipulating amygdala pathways is a potential strategy for treating cataplexy in narcolepsy.


Assuntos
Tonsila do Cerebelo/metabolismo , Cataplexia/metabolismo , Orexinas/metabolismo , Tonsila do Cerebelo/fisiologia , Animais , Cataplexia/terapia , Emoções , Feminino , Terapia Genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orexinas/genética , Sono REM
5.
Eur J Neurosci ; 44(10): 2846-2857, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27657541

RESUMO

Neurons containing melanin-concentrating hormone (MCH) are located in the hypothalamus. In mice, optogenetic activation of the MCH neurons induces both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep at night, the normal wake-active period for nocturnal rodents [R. R. Konadhode et al. (2013) J. Neurosci., 33, 10257-10263]. Here we selectively activate these neurons in rats to test the validity of the sleep network hypothesis in another species. Channelrhodopsin-2 (ChR2) driven by the MCH promoter was selectively expressed by MCH neurons after injection of rAAV-MCHp-ChR2-EYFP into the hypothalamus of Long-Evans rats. An in vitro study confirmed that the optogenetic activation of MCH neurons faithfully triggered action potentials. In the second study, in Long-Evans rats, rAAV-MCH-ChR2, or the control vector, rAAV-MCH-EYFP, were delivered into the hypothalamus. Three weeks later, baseline sleep was recorded for 48 h without optogenetic stimulation (0 Hz). Subsequently, at the start of the lights-off cycle, the MCH neurons were stimulated at 5, 10, or 30 Hz (1 mW at tip; 1 min on - 4 min off) for 24 h. Sleep was recorded during the 24-h stimulation period. Optogenetic activation of MCH neurons increased both REM and NREM sleep at night, whereas during the day cycle, only REM sleep was increased. Delta power, an indicator of sleep intensity, was also increased. In control rats without ChR2, optogenetic stimulation did not increase sleep or delta power. These results lend further support to the view that sleep-active MCH neurons contribute to drive sleep in mammals.


Assuntos
Potenciais de Ação , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/fisiologia , Melaninas/metabolismo , Neurônios/fisiologia , Hormônios Hipofisários/metabolismo , Sono REM , Ciclos de Atividade , Animais , Células Cultivadas , Ritmo Delta , Hormônios Hipotalâmicos/genética , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Melaninas/genética , Neurônios/metabolismo , Optogenética , Hormônios Hipofisários/genética , Ratos , Ratos Long-Evans
6.
J Neurosci ; 33(25): 10257-63, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23785141

RESUMO

Melanin concentrating hormone (MCH) is a cyclic neuropeptide present in the hypothalamus of all vertebrates. MCH is implicated in a number of behaviors but direct evidence is lacking. To selectively stimulate the MCH neurons the gene for the light-sensitive cation channel, channelrhodopsin-2, was inserted into the MCH neurons of wild-type mice. Three weeks later MCH neurons were stimulated for 1 min every 5 min for 24 h. A 10 Hz stimulation at the start of the night hastened sleep onset, reduced length of wake bouts by 50%, increased total time in non-REM and REM sleep at night, and increased sleep intensity during the day cycle. Sleep induction at a circadian time when all of the arousal neurons are active indicates that MCH stimulation can powerfully counteract the combined wake-promoting signal of the arousal neurons. This could be potentially useful in treatment of insomnia.


Assuntos
Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/fisiologia , Melaninas/genética , Melaninas/fisiologia , Neurônios/fisiologia , Hormônios Hipofisários/genética , Hormônios Hipofisários/fisiologia , Sono/fisiologia , Animais , Channelrhodopsins , Ritmo Circadiano/fisiologia , Cor , Ritmo Delta/fisiologia , Eletrodos Implantados , Eletroencefalografia , Hipotálamo/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Estimulação Luminosa , Plasmídeos/genética , Sono REM/fisiologia , Vigília/fisiologia
7.
Sleep ; 47(5)2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38447008

RESUMO

Dynorphin is an endogenous opiate localized in many brain regions and spinal cord, but the activity of dynorphin neurons during sleep is unknown. Dynorphin is an inhibitory neuropeptide that is coreleased with orexin, an excitatory neuropeptide. We used microendoscopy to test the hypothesis that, like orexin, the dynorphin neurons are wake-active. Dynorphin-cre mice (n = 3) were administered rAAV8-Ef1a-Con/Foff 2.0-GCaMP6M into the zona incerta-perifornical area, implanted with a GRIN lens (gradient reflective index), and electrodes to the skull that recorded sleep. One month later, a miniscope imaged calcium fluorescence in dynorphin neurons during multiple bouts of wake, non-rapid-eye movement (NREM), and rapid-eye movement (REM) sleep. Unbiased data analysis identified changes in calcium fluorescence in 64 dynorphin neurons. Most of the dynorphin neurons (72%) had the highest fluorescence during bouts of active and quiet waking compared to NREM or REM sleep; a subset (20%) were REM-max. Our results are consistent with the emerging evidence that the activity of orexin neurons can be classified as wake-max or REM-max. Since the two neuropeptides are coexpressed and coreleased, we suggest that dynorphin-cre-driven calcium sensors could increase understanding of the role of this endogenous opiate in pain and sleep.


Assuntos
Dinorfinas , Neurônios , Sono REM , Vigília , Zona Incerta , Animais , Camundongos , Dinorfinas/metabolismo , Dinorfinas/fisiologia , Neurônios/fisiologia , Orexinas/metabolismo , Orexinas/fisiologia , Sono REM/fisiologia , Vigília/fisiologia , Zona Incerta/fisiologia , Zona Incerta/fisiopatologia
8.
Sleep ; 47(3)2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-37599437

RESUMO

To determine how a waking brain falls asleep researchers have monitored and manipulated activity of neurons and glia in various brain regions. While imaging Gamma-Aminobutyric Acid (GABA) neurons in the zona incerta (ZI) we found a subgroup that anticipates onset of NREM sleep (Blanco-Centurion C, Luo S, Vidal-Ortiz A, Swank C, Shiromani PJ. Activity of a subset of vesicular GABA-transporter neurons in the ventral ZI anticipates sleep onset. Sleep. 2021;44(6):zsaa268. doi:10.1093/sleep/zsaa268.). To differentiate the GABA subtype we now image and optogenetically manipulate the ZI neurons containing the transcription factor, Lhx6. In the first study, Lhx6-cre mice (n = 5; female = 4) were given rAAV-DJ-EF1a-DIO-GCaMP6M into the ZI (isofluorane anesthesia), a GRIN lens implanted, and 21days later sleep and fluorescence in individual Lhx6 neurons were recorded for 4 hours. Calcium fluorescence was detected in 132 neurons. 45.5% of the Lhx6 neurons were REM-max; 30.3% were wake-max; 11.4% were wake + REM max; 9% were NREM-max; and 3.8% had no change. The NREM-max group of neurons fluoresced 30 seconds ahead of sleep onset. The second study tested the effects of unilateral optogenetic stimulation of the ZI Lhx6 neurons (n = 14 mice) (AAV5-Syn-FLEX-rc[ChrimsonR-tdTomato]. Stimulation at 1 and 5 Hz (1 minute on- 4 minutes off) significantly increased percent REM sleep during the 4 hours stimulation period (last half of day cycle). The typical experimental approach is to stimulate neurons in both hemispheres, but here we found that low-frequency stimulation of ZI Lhx6 neurons in one hemisphere is sufficient to shift states of consciousness. Detailed mapping combined with mechanistic testing is necessary to identify local nodes that can shift the brain between wake-sleep states.


Assuntos
Proteína Vermelha Fluorescente , Sono REM , Zona Incerta , Camundongos , Feminino , Animais , Sono REM/fisiologia , Zona Incerta/fisiologia , Optogenética , Sono/fisiologia , Neurônios , Ácido gama-Aminobutírico
9.
Sleep ; 46(3)2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36516419

RESUMO

STUDY OBJECTIVES: As in various brain regions the activity of gamma-aminobutyric acid (GABA) neurons is largely unknown, we measured in vivo changes in calcium fluorescence in GABA neurons in the zona incerta (ZI) and the ventral lateral periaqueductal grey (vlPAG), two areas that have been implicated in regulating sleep. METHODS: vGAT-Cre mice were implanted with sleep electrodes, microinjected with rAAV-DIO-GCaMP6 into the ZI (n = 6) or vlPAG (n = 5) (isoflurane anesthesia) and a GRIN (Gradient-Index) lens inserted atop the injection site. Twenty-one days later, fluorescence in individual vGAT neurons was recorded over multiple REM cycles. Regions of interest corresponding to individual vGAT somata were automatically extracted with PCA-ICA analysis. RESULTS: In the ZI, 372 neurons were identified. Previously, we had recorded the activity of 310 vGAT neurons in the ZI and we combined the published dataset with the new dataset to create a comprehensive dataset of ZI vGAT neurons (total neurons = 682; mice = 11). In the vlPAG, 169 neurons (mice = 5) were identified. In both regions, most neurons were maximally active in REM sleep (R-Max; ZI = 51.0%, vlPAG = 60.9%). The second most abundant group was W-Max (ZI = 23.9%, vlPAG = 25.4%). In the ZI, but not in vlPAG, there were neurons that were NREMS-Max (11.7%). vlPAG had REMS-Off neurons (8.3%). In both areas, there were two minor classes: wake/REMS-Max and state indifferent. In the ZI, the NREMS-Max neurons fluoresced 30 s ahead of sleep onset. CONCLUSIONS: These descriptive data show that the activity of GABA neurons is biased in favor of sleep in two brain regions implicated in sleep.


Assuntos
Zona Incerta , Camundongos , Animais , Zona Incerta/fisiologia , Substância Cinzenta Periaquedutal , Sono/fisiologia , Ácido gama-Aminobutírico , Neurônios GABAérgicos
10.
J Neurosci ; 31(16): 6028-40, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21508228

RESUMO

Cataplexy, a sudden unexpected muscle paralysis, is a debilitating symptom of the neurodegenerative sleep disorder, narcolepsy. During these attacks, the person is paralyzed, but fully conscious and aware of their surroundings. To identify potential neurons that might serve as surrogate orexin neurons to suppress such attacks, the gene for orexin (hypocretin), a peptide lost in most human narcoleptics, was delivered into the brains of the orexin-ataxin-3 transgenic mouse model of human narcolepsy. Three weeks after the recombinant adenoassociated virus (rAAV)-mediated orexin gene transfer, sleep-wake behavior was assessed. rAAV-orexin gene delivery into neurons of the zona incerta (ZI), or the lateral hypothalamus (LH) blocked cataplexy. Orexin gene transfer into the striatum or in the melanin-concentrating hormone neurons in the ZI or LH had no such effect, indicating site specificity. In transgenic mice lacking orexin neurons but given rAAV-orexin, detectable levels of orexin-A were evident in the CSF, indicating release of the peptide from the surrogate neurons. Retrograde tracer studies showed that the amygdala innervates the ZI consistent with evidence that strong emotions trigger cataplexy. In turn, the ZI projects to the locus ceruleus, indicating that the ZI is part of a circuit that stabilizes motor tone. Our results indicate that these neurons might also be recruited to block the muscle paralysis in narcolepsy.


Assuntos
Cataplexia/terapia , Terapia Genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Narcolepsia/terapia , Neurônios/metabolismo , Neuropeptídeos/genética , Subtálamo/metabolismo , Animais , Cataplexia/genética , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Técnicas de Transferência de Genes , Genótipo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Narcolepsia/genética , Neuropeptídeos/metabolismo , Orexinas , Sono
11.
Proc Natl Acad Sci U S A ; 105(29): 10227-32, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18645184

RESUMO

The presence of large-amplitude, slow waves in the EEG is a primary characteristic that distinguishes cerebral activity during sleep from that which occurs during wakefulness. Although sleep-active neurons have been identified in other brain areas, neurons that are specifically activated during slow-wave sleep have not previously been described in the cerebral cortex. We have identified a population of cells in the cortex that is activated during sleep in three mammalian species. These cortical neurons are a subset of GABAergic interneurons that express neuronal NOS (nNOS). Because Fos expression in these sleep-active, nNOS-immunoreactive (nNOS-ir) neurons parallels changes in the intensity of slow-wave activity in the EEG, and these neurons are innvervated by neurotransmitter systems previously implicated in sleep/wake control, cortical nNOS-ir neurons may be part of the neurobiological substrate that underlies homeostatic sleep regulation.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Neurônios/classificação , Neurônios/fisiologia , Sono/fisiologia , Animais , Cricetinae , Eletroencefalografia , Eletrofisiologia , Interneurônios/classificação , Interneurônios/citologia , Interneurônios/fisiologia , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Privação do Sono/patologia , Privação do Sono/fisiopatologia , Vigília/fisiologia
12.
Front Neurosci ; 15: 646468, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33828453

RESUMO

It was in the influenza pandemic of 1918 that von Economo identified specific brain regions regulating sleep and wake. Since then researchers have used a variety of tools to determine how the brain shifts between states of consciousness. In every enterprise new tools have validated existing data, corrected errors and made new discoveries to advance science. The brain is a challenge but new tools can disentangle the brain network. We summarize the newest tool, a miniature microscope, that provides unprecedented view of activity of glia and neurons in freely behaving mice. With this tool we have observed that the activity of a majority of GABA and MCH neurons in the lateral hypothalamus is heavily biased toward sleep. We suggest that miniscope data identifies activity at the cellular level in normal versus diseased brains, and also in response to specific hypnotics. Shifts in activity in small networks across the brain will help identify point of criticality that switches the brain from wake to sleep.

13.
Sleep ; 44(6)2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33270105

RESUMO

STUDY OBJECTIVES: Sleep and wake are opposing behavioral states controlled by the activity of specific neurons that need to be located and mapped. To better understand how a waking brain falls asleep it is necessary to identify activity of individual phenotype-specific neurons, especially neurons that anticipate sleep onset. In freely behaving mice, we used microendoscopy to monitor calcium (Ca2+) fluorescence in individual hypothalamic neurons expressing the vesicular GABA transporter (vGAT), a validated marker of GABA neurons. METHODS: vGAT-Cre mice (male = 3; female = 2) transfected with rAAV-FLEX-GCaMP6M in the lateral hypothalamus were imaged 30 days later during multiple episodes of waking (W), non-rapid-eye movement sleep (NREMS) or REMS (REMS). RESULTS: 372 vGAT neurons were recorded in the zona incerta. 23.9% of the vGAT neurons showed maximal fluorescence during wake (classified as wake-max), 4% were NREM-max, 56.2% REM-max, 5.9% wake/REM max, while 9.9% were state-indifferent. In the NREM-max group, Ca2+ fluorescence began to increase before onset of NREM sleep, remained high throughout NREM sleep, and declined in REM sleep. CONCLUSIONS: We found that 60.2% of the vGAT GABA neurons in the zona incerta had activity that was biased towards sleep (NREM and REMS). A subset of vGAT neurons (NREM-max) became active in advance of sleep onset and may induce sleep by inhibiting the activity of the arousal neurons. Abnormal activation of the NREM-max neurons may drive sleep attacks and hypersomnia.


Assuntos
Zona Incerta , Animais , Feminino , Masculino , Camundongos , Sono , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Vigília , Zona Incerta/metabolismo , Ácido gama-Aminobutírico
14.
J Neurosci ; 29(5): 1580-5, 2009 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19193905

RESUMO

Narcolepsy is a human sleep disorder resulting from the loss of neurons containing the neuropeptide orexin, also known as hypocretin. Cataplexy, which is a sudden loss of muscle tone during waking, is an important diagnostic symptom of narcolepsy. In humans and canines with narcolepsy, cataplexy is considered to be a separate and distinct behavioral state. However, in the mouse model of the disease this issue is not resolved. The present study monitored the activity of forty four neurons in the rostral pons in hypocretin knock-out mice. Majority of the neurons were active during wake and REM sleep, while four neurons were selectively active during REM sleep. All of these neurons were less active during cataplexy compared with REM sleep. Thus, although cataplexy and REM sleep share many common features, including the muscle atonia, cataplexy is a distinct state in mice.


Assuntos
Cataplexia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neurônios/fisiologia , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Ponte/fisiologia , Sono/genética , Animais , Cataplexia/genética , Eletroencefalografia/métodos , Camundongos , Camundongos Knockout , Orexinas , Ponte/citologia , Sono/fisiologia , Sono REM/genética , Sono REM/fisiologia
15.
IBRO Rep ; 6: 132-136, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31193158

RESUMO

The amygdala regulates multiple behaviors and emotions by projecting to multiple brain regions. However, the topographical distribution of amygdala neurons projecting to specific brain areas is still unclear. In the present study, we focus on determining whether single amygdala neurons project to the brain stem ventrolateral periaqueductal grey (vlPAG) and to the medial prefrontal cortex (mPFC). The mPFC neurons are involved in detecting emotional content while the vlPAG neurons are involved in regulating muscle tone. In VGAT-Cre mice a cre-inducible retrograde AAV tracer expressing tdTomato was microinjected into the ventrolateral periaqueductal grey matter (vlPAG), while a second retrograde AAV tracer with generic expression of GFP was delivered into the medial prefrontal cortex (mPFC). The results identified a subgroup of bifurcating GABAergic neurons in the central nucleus (CeA) and basolateral amygdala (BLA) that projects to vlPAG and mPFC. Based on these projections we suggest that amygdala GABA neurons may be involved in triggering emotionally-induced cataplexy in the sleep disorder, narcolepsy.

16.
J Neurosci ; 27(51): 14041-8, 2007 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18094243

RESUMO

The hypocretin (HCRT) neurons are located only in the perifornical area of the lateral hypothalamus and heavily innervate the cholinergic neurons in the basal forebrain (BF), histamine neurons in the tuberomammillary nucleus (TMN), and the noradrenergic locus ceruleus (LC) neurons, three neuronal populations that have traditionally been implicated in regulating arousal. Based on the innervation, HCRT neurons may regulate arousal by driving these downstream arousal neurons. Here, we directly test this hypothesis by a simultaneous triple lesion of these neurons using three saporin-conjugated neurotoxins. Three weeks after lesion, the daily levels of wake were not changed in rats with double or triple lesions, although rats with triple lesions were asleep more during the light-to-dark transition period. The double- and triple-lesioned rats also had more stable sleep architecture compared with nonlesioned saline rats. These results suggest that the cholinergic BF, TMN, and LC neurons jointly modulate arousal at a specific circadian time, but they are not essential links in the circuitry responsible for daily levels of wake, as traditionally hypothesized.


Assuntos
Nível de Alerta/fisiologia , Neurônios/fisiologia , Periodicidade , Proteínas de Plantas/toxicidade , Proteínas Inativadoras de Ribossomos Tipo 1/toxicidade , Sono/fisiologia , Animais , Nível de Alerta/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Neuropeptídeos/fisiologia , Orexinas , Ratos , Ratos Sprague-Dawley , Saporinas , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , Vigília/fisiologia
17.
Eur J Neurosci ; 28(6): 1191-8, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18783368

RESUMO

Neurons containing the neuropeptide hypocretin (HCRT, orexin) are localized only in the lateral hypothalamus, from where they innervate multiple regions implicated in arousal, including the basal forebrain. HCRT activation of downstream arousal neurons is likely to stimulate release of endogenous factors. One such factor is adenosine, which in the basal forebrain increases in level with wakefulness and decreases with sleep, and is hypothesized to regulate the waxing and waning of sleep drive. Does loss of HCRT neurons affect adenosine levels in the basal forebrain? Is the increased sleep that accompanies HCRT loss a consequence of higher adenosine levels in the basal forebrain? In the present study, we investigated these questions by lesioning the HCRT neurons with HCRT-2-saporin (HCRT-2-SAP) and measuring sleep and extracellular levels of adenosine in the basal forebrain. In separate groups of rats, the neurotoxin HCRT-2-SAP or saline was administered locally to the lateral hypothalamus, and 80 days later adenosine and sleep were assessed. Rats given the neurotoxin had a 94% loss of HCRT neurons. These rats woke less at night, and had more rapid eye movement sleep, which is consistent with HCRT hypofunction. These rats also had more sleep after brief periods of sleep deprivation. However, in the lesioned rats, adenosine levels did not increase with 6 h of sleep deprivation, whereas an increase in adenosine levels occurred in rats without lesion of the HCRT neurons. These findings indicate that adenosine levels do not increase with wakefulness in rats with a HCRT lesion, and that the increased sleep in these rats occurs independently of adenosine levels in the basal forebrain.


Assuntos
Adenosina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Prosencéfalo/metabolismo , Sono/fisiologia , Animais , Masculino , Microdiálise , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/farmacologia , Neuropeptídeos/toxicidade , Orexinas , Prosencéfalo/citologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/patologia , Ratos , Ratos Sprague-Dawley , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Proteínas Inativadoras de Ribossomos Tipo 1/toxicidade , Saporinas , Privação do Sono/metabolismo , Toxinas Biológicas/farmacologia , Toxinas Biológicas/toxicidade , Vigília/fisiologia
18.
Eur J Neurosci ; 28(7): 1382-93, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18973565

RESUMO

Gene transfer has proven to be an effective neurobiological tool in a number of neurodegenerative diseases, but it is not known if it can correct a sleep disorder. Narcolepsy is a neurodegenerative sleep disorder linked to the loss of neurons containing the neuropeptide orexin, also known as hypocretin. Here, a replication-defective herpes simplex virus-1 amplicon-based vector was constructed to transfer the gene for mouse prepro-orexin into mice with a genetic deletion of the orexin gene. After in vitro tests confirmed successful gene transfer into cells, the gene vector was delivered to the lateral hypothalamus of orexin knockout (KO) mice where the orexin peptide was robustly expressed in the somata and processes of numerous neurons, and the peptide product was detected in the cerebrospinal fluid. During the 4-day life-span of the vector the incidence of cataplexy declined by 60%, and the levels of rapid eye movement sleep during the second half of the night were similar to levels in wild-type mice, indicating that narcoleptic sleep-wake behavior in orexin KO mice can be improved by targeted gene transfer.


Assuntos
Técnicas de Transferência de Genes , Região Hipotalâmica Lateral/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Narcolepsia/genética , Narcolepsia/metabolismo , Neuropeptídeos/genética , Sono/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Genes Reporter/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/cirurgia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Narcolepsia/terapia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Resultado do Tratamento
19.
Brain Res ; 1205: 47-54, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18343358

RESUMO

Ablation of the SCN, an established circadian clock, does not abolish food entrainment, suggesting that the food-entrainable oscillator (FEO) must lie outside the SCN. Typically, animals show anticipatory locomotor activity and rise in core body temperature under the influence of the FEO. Signals from the FEO would, therefore, converge onto arousal neurons so that the animal might forage for food. In the present study, we investigate whether the neuropeptide orexin, which has been linked to arousal, might transduce the arousal signal. Orexin-knockout (orexin-KO) and wildtype (WT) mice (both C57BL/6J derived) were implanted with MiniMitter transmitters that recorded core body temperature and activity (12 h LD cycle). After a week of ad-libitum feeding, the mice were given access to food for 4 h (ZT 4-8) for nine days followed by 2-days of fasting. When orexin-KO mice were placed in a restricted feeding schedule, both core body temperature and activity entrained to the feeding schedule. In these mice gross locomotor activity was severely blunted during the nine day period of restricted feeding (-79.4+/-6.3%) from the WT, but they showed an increase in core body temperature in anticipation to the meal time similar to the WT mice. There was no difference in the amount of food intake between the genotypes. We conclude that orexin is not required for entrainment of activity and temperature to a restricted feeding schedule, but is required for the robust expression of gross locomotor activity in anticipation of the scheduled feeding.


Assuntos
Restrição Calórica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Atividade Motora/fisiologia , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Periodicidade , Temperatura , Animais , Nível de Alerta/fisiologia , Jejum/fisiologia , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orexinas , Transdução de Sinais/fisiologia
20.
IBRO Rep ; 4: 44-49, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30155524

RESUMO

The neuropeptides orexin and melanin-concentrating hormone (MCH), as well as the neurotransmitters GABA (γ-Aminobutyric acid) and glutamate are chief modulators of the sleep-wake states in the posterior hypothalamus. To investigate co-expression of vesicular GABA transporter (VGAT, a marker of GABA neurons) and the vesicular glutamate transporter-2 (VGLUT2, a marker of glutamate neurons) in orexin and MCH neurons, we generated two transgenic mouse lines. One line selectively expressed the reporter gene EYFP in VGAT+ neurons, whereas the other line expressed reporter gene tdTomato in VGLUT2+ neurons. Co-localization between these genetic reporters and orexin or MCH immunofluorescent tags was determined using 3D computer reconstructions of Z stacks that were acquired using a multiphoton laser confocal microscope. Our results demonstrated that MCH neurons expressed neither VGAT nor VGLUT2, suggesting MCH neurons are a separate cluster of cells from VGAT+ GABAergic neurons and VGLUT2+ glutamatergic neurons. Moreover, most orexin neurons expressed VGLUT2, indicating these neurons are glutamatergic. Our data suggested that in the posterior hypothalamus there are four major distinct groups of neurons: VGAT+, orexin+/VGLUT2+, orexin-/VGLUT2+, and MCH neurons. This study facilitated our understanding of the role of these neurotransmitters and neuropeptides in relation to sleep/wake regulation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA